scispace - formally typeset
Search or ask a question

Showing papers on "Dosage compensation published in 2010"


Journal ArticleDOI
TL;DR: This work reviews the main mechanisms responsible for increased immune activity in females, which provides a survival advantage in the face of pathogenic insult but can also enhance the susceptibility of females to autoimmunity.
Abstract: In response to various immune challenges, females show better survival than males; the X chromosome has an important role in this immunological advantage. X chromosome-linked diseases are usually restricted to males, who have only one copy of the X chromosome; however, females are more prone to autoimmune diseases, and the X chromosome may be involved in the breakdown of self tolerance. Several hypotheses have been proposed in recent years that support a role for the X chromosome in shaping autoimmune responses. Here, we review the main mechanisms responsible for increased immune activity in females. This provides a survival advantage in the face of pathogenic insult but can also enhance the susceptibility of females to autoimmunity.

582 citations


Journal ArticleDOI
01 Feb 2010-Fly
TL;DR: Recent advances in the understanding of Sxl regulation and function are described, highlighting work that challenges some of the textbook views about this classical (often cited, yet poorly understood) binary switch gene.
Abstract: One of the most important decisions in development is whether to be male or female. In Drosophila melanogaster, most cells make this choice independent of their neighbors such that diploid cells with one X chromosome (XY) are male and those with two X chromosomes (XX) are female. X-chromosome number is relayed through regulatory proteins that act together to activate Sex-lethal (Sxl) in XX animals. The resulting SXL female specific RNA binding protein modulates the expression of a set of downstream genes, ultimately leading to sexually dimorphic structures and behaviors. Despite the apparent simplicity of this mechanism, Sxl activity is controlled by a host of transcriptional and posttranscriptional mechanisms that tailor its function to specific developmental scenarios. This review describes recent advances in our understanding of Sxl regulation and function, highlighting work that challenges some of the textbook views about this classical (often cited, yet poorly understood) binary switch gene.

239 citations


Journal ArticleDOI
TL;DR: Structural analyses provide new insights into the folding of the A region of the Xist RNA, which plays a crucial role in X chromosome inactivation, and its mechanism of protein recruitment.
Abstract: In placental mammals, inactivation of one of the X chromosomes in female cells ensures sex chromosome dosage compensation. The 17 kb non-coding Xist RNA is crucial to this process and accumulates on the future inactive X chromosome. The most conserved Xist RNA region, the A region, contains eight or nine repeats separated by U-rich spacers. It is implicated in the recruitment of late inactivated X genes to the silencing compartment and likely in the recruitment of complex PRC2. Little is known about the structure of the A region and more generally about Xist RNA structure. Knowledge of its structure is restricted to an NMR study of a single A repeat element. Our study is the first experimental analysis of the structure of the entire A region in solution. By the use of chemical and enzymatic probes and FRET experiments, using oligonucleotides carrying fluorescent dyes, we resolved problems linked to sequence redundancies and established a 2-D structure for the A region that contains two long stem-loop structures each including four repeats. Interactions formed between repeats and between repeats and spacers stabilize these structures. Conservation of the spacer terminal sequences allows formation of such structures in all sequenced Xist RNAs. By combination of RNP affinity chromatography, immunoprecipitation assays, mass spectrometry, and Western blot analysis, we demonstrate that the A region can associate with components of the PRC2 complex in mouse ES cell nuclear extracts. Whilst a single four-repeat motif is able to associate with components of this complex, recruitment of Suz12 is clearly more efficient when the entire A region is present. Our data with their emphasis on the importance of inter-repeat pairing change fundamentally our conception of the 2-D structure of the A region of Xist RNA and support its possible implication in recruitment of the PRC2 complex.

236 citations


Journal ArticleDOI
29 Jul 2010-Nature
TL;DR: It is concluded that the avian Z and mammalian X chromosomes followed convergent evolutionary trajectories, despite their evolving with opposite (female versus male) systems of heterogamety.
Abstract: Birds and mammals have distinct sex chromosomes. In birds, males have a pair of Z chromosomes and females a Z and a W. In mammals, males are XY and females XX. It has long been assumed that sex-chromosome evolution has involved dramatic modification of the sex-specific (W and Y) chromosomes but only modest changes to the Z and X chromosomes shared by the sexes. Not so, according to a new study reporting the sequence of the chicken Z chromosome and comparing it with the finished sequence of human X. The Z and X chromosomes have changed dramatically from the autosomal (non-sex) chromosomes that gave rise to them. And they seem to have followed convergent evolutionary trajectories, including the acquisition and amplification of testis-expressed gene families, despite having arisen independently from different portions of the ancestral genome. Birds and mammals have distinct sex chromosomes: in birds, males are ZZ and females ZW; in mammals, males are XY and females XX. By sequencing the chicken Z chromosome and comparing it with the human X chromosome, these authors overturn the currently held view that these chromosomes have diverged little from their autosomal progenitors. The Z and X chromosomes seem to have followed convergent evolutionary trajectories, despite evolving with opposite systems of heterogamety. In birds, as in mammals, one pair of chromosomes differs between the sexes. In birds, males are ZZ and females ZW. In mammals, males are XY and females XX. Like the mammalian XY pair, the avian ZW pair is believed to have evolved from autosomes, with most change occurring in the chromosomes found in only one sex—the W and Y chromosomes1,2,3,4,5. By contrast, the sex chromosomes found in both sexes—the Z and X chromosomes—are assumed to have diverged little from their autosomal progenitors2. Here we report findings that challenge this assumption for both the chicken Z chromosome and the human X chromosome. The chicken Z chromosome, which we sequenced essentially to completion, is less gene-dense than chicken autosomes but contains a massive tandem array containing hundreds of duplicated genes expressed in testes. A comprehensive comparison of the chicken Z chromosome with the finished sequence of the human X chromosome demonstrates that each evolved independently from different portions of the ancestral genome. Despite this independence, the chicken Z and human X chromosomes share features that distinguish them from autosomes: the acquisition and amplification of testis-expressed genes, and a low gene density resulting from an expansion of intergenic regions. These features were not present on the autosomes from which the Z and X chromosomes originated but were instead acquired during the evolution of Z and X as sex chromosomes. We conclude that the avian Z and mammalian X chromosomes followed convergent evolutionary trajectories, despite their evolving with opposite (female versus male) systems of heterogamety. More broadly, in birds and mammals, sex chromosome evolution involved not only gene loss in sex-specific chromosomes, but also marked expansion and gene acquisition in sex chromosomes common to males and females.

206 citations


Journal ArticleDOI
TL;DR: It is shown that RNA sequencing is more sensitive than microarray and that RNA-Seq data reveal an X:AA ratio of ∼0.5 in human and mouse, necessitating a major revision of the current model of dosage compensation in the evolution of sex chromosomes.
Abstract: Jianzhi Zhang and Xionglei He report analyses of published RNA sequencing data examining relative expression levels between genes located on the X chromosome and genes located on autosomes. Unlike previous reports of dosage compensation between the X chromosome and autosomes, their analyses detect an X:autosome expression ratio of ∼0.5.

191 citations


Journal ArticleDOI
TL;DR: Two bursts of gene gains occurred on the mammalian X chromosome contribute to an age-dependent chromosomal distribution of male-biased genes.
Abstract: Mammalian X chromosomes evolved under various mechanisms including sexual antagonism, the faster-X process, and meiotic sex chromosome inactivation (MSCI). These forces may contribute to nonrandom chromosomal distribution of sex-biased genes. In order to understand the evolution of gene content on the X chromosome and autosome under these forces, we dated human and mouse protein-coding genes and miRNA genes on the vertebrate phylogenetic tree. We found that the X chromosome recently acquired a burst of young male-biased genes, which is consistent with fixation of recessive male-beneficial alleles by sexual antagonism. For genes originating earlier, however, this pattern diminishes and finally reverses with an overrepresentation of the oldest male-biased genes on autosomes. MSCI contributes to this dynamic since it silences X-linked old genes but not X-linked young genes. This demasculinization process seems to be associated with feminization of the X chromosome with more X-linked old genes expressed in ovaries. Moreover, we detected another burst of gene originations after the split of eutherian mammals and opossum, and these genes were quickly incorporated into transcriptional networks of multiple tissues. Preexisting X-linked genes also show significantly higher protein-level evolution during this period compared to autosomal genes, suggesting positive selection accompanied the early evolution of mammalian X chromosomes. These two findings cast new light on the evolutionary history of the mammalian X chromosome in terms of gene gain, sequence, and expressional evolution.

190 citations


Journal ArticleDOI
TL;DR: Analysis of the relationship between gene copy number and gene expression in aneuploid male Drosophila cells reveals a global compensation mechanism in addition to X chromosome-specific dosage compensation.
Abstract: Extensive departures from balanced gene dose in aneuploids are highly deleterious However, we know very little about the relationship between gene copy number and expression in aneuploid cells We determined copy number and transcript abundance (expression) genome-wide in Drosophila S2 cells by DNA-Seq and RNA-Seq We found that S2 cells are aneuploid for >43 Mb of the genome, primarily in the range of one to five copies, and show a male genotype (∼ two X chromosomes and four sets of autosomes, or 2X;4A) Both X chromosomes and autosomes showed expression dosage compensation X chromosome expression was elevated in a fixed-fold manner regardless of actual gene dose In engineering terms, the system “anticipates” the perturbation caused by X dose, rather than responding to an error caused by the perturbation This feed-forward regulation resulted in precise dosage compensation only when X dose was half of the autosome dose Insufficient compensation occurred at lower X chromosome dose and excessive expression occurred at higher doses RNAi knockdown of the Male Specific Lethal complex abolished feed-forward regulation Both autosome and X chromosome genes show Male Specific Lethal–independent compensation that fits a first order dose-response curve Our data indicate that expression dosage compensation dampens the effect of altered DNA copy number genome-wide For the X chromosome, compensation includes fixed and dose-dependent components

166 citations


Journal ArticleDOI
TL;DR: The vast diversity of sex chromosome systems in insects--ranging from the classical male heterogametic XY system in Drosophila to ZW systems in Lepidoptera or mobile genes determining sex as found in house flies--implies that insects can serve as unique model systems to study various functional and evolutionary aspects of these different processes.
Abstract: Sex chromosomes have many unusual features relative to autosomes. Y (or W) chromosomes lack genetic recombination, are male- (female-) limited, and show an abundance of genetically inert heterochromatic DNA but contain few functional genes. X (or Z) chromosomes also show sex-biased transmission (i.e., X chromosomes show female-biased and Z-chromosomes show male-biased inheritance) and are hemizygous in the heterogametic sex. Their unusual ploidy level and pattern of inheritance imply that sex chromosomes play a unique role in many biological processes and phenomena, including sex determination, epigenetic chromosome-wide regulation of gene expression, the distribution of genes in the genome, genomic conflict, local adaptation, and speciation. The vast diversity of sex chromosome systems in insects—ranging from the classical male heterogametic XY system in Drosophila to ZW systems in Lepidoptera or mobile genes determining sex as found in house flies—implies that insects can serve as unique model systems t...

131 citations


Journal ArticleDOI
04 Jun 2010-PLOS ONE
TL;DR: It is shown that XCI is random in human placenta, and that this organ is arranged in relatively large patches of cells with either maternal or paternal inactive X, which combined with the extensive mosaicism found in placente can explain the lack of agreement among previous studies.
Abstract: Imprinted inactivation of the paternal X chromosome in marsupials is the primordial mechanism of dosage compensation for X-linked genes between females and males in Therians. In Eutherian mammals, X chromosome inactivation (XCI) evolved into a random process in cells from the embryo proper, where either the maternal or paternal X can be inactivated. However, species like mouse and bovine maintained imprinted XCI exclusively in extraembryonic tissues. The existence of imprinted XCI in humans remains controversial, with studies based on the analyses of only one or two X-linked genes in different extraembryonic tissues. Here we readdress this issue in human term placenta by performing a robust analysis of allele-specific expression of 22 X-linked genes, including XIST, using 27 SNPs in transcribed regions. We show that XCI is random in human placenta, and that this organ is arranged in relatively large patches of cells with either maternal or paternal inactive X. In addition, this analysis indicated heterogeneous maintenance of gene silencing along the inactive X, which combined with the extensive mosaicism found in placenta, can explain the lack of agreement among previous studies. Our results illustrate the differences of XCI mechanism between humans and mice, and highlight the importance of addressing the issue of imprinted XCI in other species in order to understand the evolution of dosage compensation in placental mammals.

130 citations


Journal ArticleDOI
TL;DR: It is found that the imprinted XP can be functionally divided into two domains, one comprising traditional coding genes (genic) and the other comprising intergenic repetitive elements, which suggests that Xist RNA functions relatively late during preimplantation mouse development.
Abstract: Genomic imprinting refers to a parent-of-origin effect on gene expression in the developing embryo (3, 57). The existence of imprinting in the mammal means that male and female gametes contribute significantly different information to the zygote. One important difference is illustrated by X chromosome inactivation (XCI), the mechanism of dosage compensation in the mammal that results in the silencing of one X chromosome in the female embryo (2, 33, 34, 49, 64). While the eutherian form of XCI occurs randomly in the soma, the marsupial form is imprinted to occur exclusively on the paternal X (XP) (54). Imprinted XCI also occurs in some eutherians but is restricted to the preimplantation embryo and the extraembryonic tissues (25, 37, 47, 60). Imprinted XCI precedes random XCI in the early mouse embryo and continues through the placental lineages. In the epiblast (embryo proper), transient X reactivation is followed by random XCI, which accounts for the mosaic pattern of inactivation seen in all somatic tissues of the eutherian. The mechanisms and developmental timing of imprinted XCI remain unclear and are much debated. In principle, the maternal or paternal germ line (or both) may differentially mark the X chromosomes, with the maternal imprint protecting the maternal X (XM) from inactivation and/or the paternal mark predestining XP for inactivation. The search for parent-specific regulators frequently has focused on the X inactivation center (Xic) (7), an X-linked region harboring several noncoding regulators for random XCI. Xist produces a 17-kb noncoding transcript whose accumulation on the X has been associated with the initiation of both random and imprinted XCI (6, 38, 50). In the preimplantation mouse embryo and in the extraembryonic lineages of the postimplantation embryo, Xist is imprinted to be paternally expressed in accordance with preferential XP inactivation (28). The randomization of Xist expression following X reactivation in the epiblast lineage results in mosaic XM and XP inactivation in the embryo proper. The Xic also harbors Tsix, the 40-kb noncoding transcript that is complementary to and negatively regulates Xist (31). In contrast to Xist, Tsix is imprinted to be maternally expressed and therefore may be a maternal factor that protects XM from silencing in the early embryo and extraembryonic tissues (30, 53). Tsix expression also becomes randomized following X reactivation in the epiblast and is expressed exclusively from the future active X (Xa) in the developing embryo proper. In the eutherian embryo, the importance of Xist/Tsix in the imprinting of the X has been borne out by genetic analyses: deleting Xist from XP causes a loss of XP silencing in the placental lineages (38), whereas deleting Xist from XM (on which it is normally silent) has no consequence; conversely, deleting Tsix from XP (on which it is normally silent) has no consequence, whereas deleting Tsix from XM results in ectopic XCI on XM in the placental lineages (30, 53). Thus, for both imprinted and random XCI, Xist designates the future inactive X (Xi), while Tsix designates the future active X (Xa). Although Xic clearly regulates imprinting in eutherians, XIC or an equivalent has yet to be identified in marsupials (11, 12, 22, 55). The absence of a marsupial Xist suggests that an alternative means of silencing the X must occur in mammals. Since the discovery of meiotic sex chromosome inactivation (MSCI) in the male germ line of both eutherian and marsupial mammals (14, 23, 32, 43, 58), several groups have hypothesized a link between MSCI and the imprinting of XP (10, 24, 26, 35, 39). Recent reports that XY silencing persists into the long postmeiotic period of spermatogenesis (16, 42, 62) support the idea that zygotic XP silencing is built in part on MSCI and its aftereffects in the paternal germ line. Because MSCI is Xist independent and Xist is not highly expressed during spermatogenesis (40, 61), germ line-driven silencing would provide an alternative imprinting mechanism that would not require an XIC in the marsupial and would dosage compensate the marsupial zygote from the time of conception. The probability of an XIST-independent mechanism in the marsupial raises intriguing questions for imprinted XCI in eutherians. Did eutherian XCI evolve completely independently, or do vestiges of a marsupial mechanism still exist in the eutherians of today? Although the placental form of imprinted XCI in the mouse clearly depends on Xist (38), the role of Xist in the preimplantation embryo currently is unclear. Indeed, embryos deleted for Xist on XP are normal in the preimplantation stages and perish only after uterine implantation and the outgrowth of a placenta (38), suggesting that the early mouse embryo does not require Xist. There also is debate as to whether mouse XP is inherited from the male germ line in a partially inactive state, further raising the question of whether Xist is required to initiate imprinted XCI in the early mouse embryo (25, 45, 46). Here, we investigate the mechanism of XP silencing in the earliest stages following the gamete-to-embryo transition. We discover that imprinted XP silencing takes place in two sequential steps, one involving repetitive elements and the other involving coding genes, and implicate repeats in the transmission of parental information to the early embryo.

126 citations


Journal ArticleDOI
TL;DR: This work has clarified the molecular nature of the Caenorhabditis elegans sex-determination signal, which tallies X-chromosome number relative to the ploidy and controls both the choice of sexual fate and the process of dosage compensation.

Journal ArticleDOI
TL;DR: The zebra finch differs from the chicken because it lacks a specialized region of greater dosage compensation along the Z chromosome, and shows other differences in sex bias.
Abstract: We compared global patterns of gene expression between two bird species, the chicken and zebra finch, with regard to sex bias of autosomal versus Z chromosome genes, dosage compensation, and evolution of sex bias. Both species appear to lack a Z chromosome-wide mechanism of dosage compensation, because both have a similar pattern of significantly higher expression of Z genes in males relative to females. Unlike the chicken Z chromosome, which has female-specific expression of the noncoding RNA MHM (male hypermethylated) and acetylation of histone 4 lysine 16 (H4K16) near MHM, the zebra finch Z chromosome appears to lack the MHM sequence and acetylation of H4K16. The zebra finch also does not show the reduced male-to-female (M:F) ratio of gene expression near MHM similar to that found in the chicken. Although the M:F ratios of Z chromosome gene expression are similar across tissues and ages within each species, they differ between the two species. Z genes showing the greatest species difference in M:F ratio were concentrated near the MHM region of the chicken Z chromosome. This study shows that the zebra finch differs from the chicken because it lacks a specialized region of greater dosage compensation along the Z chromosome, and shows other differences in sex bias. These patterns suggest that different avian taxa may have evolved specific compensatory mechanisms.

Journal ArticleDOI
TL;DR: The study suggests that chromosomal translocations can result in hitherto unforeseen, large-scale changes in gene expression that are the consequence of alterations in normal chromosome territory positioning, which has consequences for the patterns of gene expression change seen during tumorigenesis-associated genome instability and during the karyotype changes that lead to speciation.
Abstract: Translocations are known to affect the expression of genes at the breakpoints and, in the case of unbalanced translocations, alter the gene copy number. However, a comprehensive understanding of the functional impact of this class of variation is lacking. Here, we have studied the effect of balanced chromosomal rearrangements on gene expression by comparing the transcriptomes of cell lines from controls and individuals with the t(11;22)(q23;q11) translocation. The number of differentially expressed transcripts between translocation-carrying and control cohorts is significantly higher than that observed between control samples alone, suggesting that balanced rearrangements have a greater effect on gene expression than normal variation. Many of the affected genes are located along the length of the derived chromosome 11. We show that this chromosome is concomitantly altered in its spatial organization, occupying a more central position in the nucleus than its nonrearranged counterpart. Derivative 22-mapping chromosome 22 genes, on the other hand, remain in their usual environment. Our results are consistent with recent studies that experimentally altered nuclear organization, and indicated that nuclear position plays a functional role in regulating the expression of some genes in mammalian cells. Our study suggests that chromosomal translocations can result in hitherto unforeseen, large-scale changes in gene expression that are the consequence of alterations in normal chromosome territory positioning. This has consequences for the patterns of gene expression change seen during tumorigenesis-associated genome instability and during the karyotype changes that lead to speciation.

Journal ArticleDOI
10 Mar 2010-PLOS ONE
TL;DR: It is shown that CNVs can directly affect expression levels of contained genes and that complexes and other dosage sensitive genes are underrepresented in CNV regions, with a particular bias against frequent variations and duplications.
Abstract: Dosage sensitivity is an important evolutionary force which impacts on gene dispensability and duplicability. The newly available data on human copy-number variation (CNV) allow an analysis of the most recent and ongoing evolution. Provided that heterozygous gene deletions and duplications actually change gene dosage, we expect to observe negative selection against CNVs encompassing dosage sensitive genes. In this study, we make use of several sources of population genetic data to identify selection on structural variations of dosage sensitive genes. We show that CNVs can directly affect expression levels of contained genes. We find that genes encoding members of protein complexes exhibit limited expression variation and overlap significantly with a manually derived set of dosage sensitive genes. We show that complexes and other dosage sensitive genes are underrepresented in CNV regions, with a particular bias against frequent variations and duplications. These results suggest that dosage sensitivity is a significant force of negative selection on regions of copy-number variation.

Journal ArticleDOI
TL;DR: The first whole-genome study of sex-biased expression in the red flour beetle, Tribolium castaneum, is presented and it is proposed that the overexpression of X chromosomes in females is an evolutionary side effect of the need to dosage compensate in males and that mechanisms to reduce female X chromosome gene expression to autosomal levels are sufficient but imperfect.
Abstract: A genome’s ability to produce two separate sexually dimorphic phenotypes is an intriguing biological mystery. Microarraybased studies of a handful of model systems suggest that much of the mystery can be explained by sex-biased gene expression evolved in response to sexually antagonistic selection. We present the first whole-genome study of sex-biased expression in the red flour beetle, Tribolium castaneum. Tribolium is a model for the largest eukaryotic order, Coleoptera, and we show that in whole-body adults, ;20% of the transcriptome is differentially regulated between the sexes. Among T. castaneum, Drosophila melanogaster, and Anopheles gambiae, we identify 416 1:1:1 orthologs with conserved sex-biased expression. Overrepresented functional categories among sex-biased genes are primarily those involved in gamete production and development. The genomic distribution of sex-biased genes in T. castaneum is distinctly nonrandom, with the strongest deficit of male-biased genes on the X chromosome (9 of 793) of any species studied to date. Tribolium also shows a significant enrichment of X-linked female-biased genes (408 of 793). Our analyses suggest that the extensive female bias of Tribolium X chromosome gene expression is due to hyperexpression of X-linked genes in both males and females. We propose that the overexpression of X chromosomes in females is an evolutionary side effect of the need to dosage compensate in males and that mechanisms to reduce female X chromosome gene expression to autosomal levels are sufficient but imperfect.

Journal ArticleDOI
TL;DR: It is hypothesized that degeneration of the Y chromosome results in regulatory mutations that create a sex-specific expression pattern and that this physical concentration of sex-biased expression on the nascent sex chromosome may be a key feature characterizing intermediate phases of sex chromosome evolution.
Abstract: Given that the genome of males and females are almost identical with the exception of genes on the Y (or W) chromosome or sex-determining alleles (in organisms without sex chromosomes), it is likely that many downstream processes resulting in sexual dimorphism are produced by changes in regulation. In early stages of sex chromosome evolution, as the Y-chromosome degenerates, gene expression should be significantly impacted for genes residing on the sex chromosome pair as regulatory mutations accumulate. However, this has rarely been examined because most model organisms have clearly diverged sex chromosomes. Fish provide a unique opportunity to examine the evolution of sex chromosomes because genetic sex determination has evolved quite recently in some groups of fish. We compared sex-specific transcription in threespine stickleback (Gasterosteus aculeatus) liver tissue using a long-oligo microarray. Of the 1,268 genes that were differentially expressed between sexes, a highly significant proportion (23%) was concentrated on chromosome 19, corresponding to the recently described nascent sex chromosomes. The sex-biased genes are enriched for different functional categories in males and females, although there is no specific functional enrichment on the sex chromosomes. Female-biased genes are concentrated at one end of the sex chromosome, corresponding to a deletion in the Y, suggesting a lack of global dosage compensation. Prior research on threespine sticklebacks has demonstrated various degrees of dissimilarity in upstream regions of genes on the Y providing a potential mechanism for the observed patterns of female-biased expression. We hypothesize that degeneration of the Y chromosome results in regulatory mutations that create a sex-specific expression pattern and that this physical concentration of sex-biased expression on the nascent sex chromosome may be a key feature characterizing intermediate phases of sex chromosome evolution.

Journal ArticleDOI
TL;DR: An assay for estimating relative expression per cell is described and this experimental approach will add value to transcript profiling experiments involving interspecies and interploidy comparisons by converting expression per transcriptome to expression per genome, eliminating the need for assumptions about transcriptome size.
Abstract: Evolutionary biologists are increasingly comparing gene expression patterns across species. Due to the way in which expression assays are normalized, such studies provide no direct information about expression per gene copy (dosage responses) or per cell and can give a misleading picture of genes that are differentially expressed. We describe an assay for estimating relative expression per cell. When used in conjunction with transcript profiling data, it is possible to compare the sizes of whole transcriptomes, which in turn makes it possible to compare expression per cell for each gene in the transcript profiling data set. We applied this approach, using quantitative reverse transcriptase-polymerase chain reaction and high throughput RNA sequencing, to a recently formed allopolyploid and showed that its leaf transcriptome was approximately 1.4-fold larger than either progenitor transcriptome (70% of the sum of the progenitor transcriptomes). In contrast, the allopolyploid genome is 94.3% as large as the sum of its progenitor genomes and retains ≥93.5% of the sum of its progenitor gene complements. Thus, “transcriptome downsizing” is greater than genome downsizing. Using this transcriptome size estimate, we inferred dosage responses for several thousand genes and showed that the majority exhibit partial dosage compensation. Homoeologue silencing is nonrandomly distributed across dosage responses, with genes showing extreme responses in either direction significantly more likely to have a silent homoeologue. This experimental approach will add value to transcript profiling experiments involving interspecies and interploidy comparisons by converting expression per transcriptome to expression per genome, eliminating the need for assumptions about transcriptome size.

Journal ArticleDOI
TL;DR: It is shown that dosage compensation (DC) in Drosophila may contribute substantially to the depletion of male genes on the X, both by limiting the emergence of male-biased expression patterns of existing X genes, and by contributing to gene trafficking ofmale genes off the X.

Journal ArticleDOI
01 Feb 2010-Heredity
TL;DR: The results are the first report of dosage effect on gene transcription in a vertebrate, and they support the observation that diploid and triploid salmon are generally phenotypically indistinguishable, except under stress, when triploids show reduced performance.
Abstract: Understanding how organisms function at the level of gene expression is becoming increasingly important for both ecological and evolutionary studies. It is evident that the diversity and complexity of organisms are not dependent solely on their number of genes, but also the variability in gene expression and gene interactions. Furthermore, slight differences in transcription control can fundamentally affect the fitness of the organism in a variable environment or during development. In this study, triploid and diploid Chinook salmon (Oncorhynchus tshawytscha) were used to examine the effects of polyploidy on specific and genome-wide gene expression response using quantitative real-time PCR (qRT-PCR) and microarray technology after an immune challenge with the pathogen Vibrio anguillarum. Although triploid and diploid fish had significant differences in mortality, qRT-PCR revealed no differences in cytokine gene expression response (interleukin-8, interleukin-1, interleukin-8 receptor and tumor necrosis factor), whereas differences were observed in constitutively expressed genes, (immunoglobulin (Ig) M, major histocompatibility complex (MHC) -II and β-actin) upon live Vibrio anguillarum exposure. Genome-wide microarray analysis revealed that, overall, triploid gene expression is similar to diploids, consistent with their similar phenotypes. This pattern, however, can subtly be altered under stress (for example, handling, V. anguillarum challenge) as we have observed at some housekeeping genes. Our results are the first report of dosage effect on gene transcription in a vertebrate, and they support the observation that diploid and triploid salmon are generally phenotypically indistinguishable, except under stress, when triploids show reduced performance.

Journal ArticleDOI
TL;DR: The reconstitution of this phenomenon at a model locus revealed that the activation potential of MOF is constrained in male cells in the context of the DCC to arrive at the 2-fold activation of transcription characteristic of dosage compensation.

Journal ArticleDOI
TL;DR: It is suggested that female marsupials use a repressive histone-mediated inactivation mechanism and that this may represent an ancestral dosage compensation process that differs from eutherians that require Xist transcription and DNA methylation.
Abstract: X chromosome dosage compensation in female eutherian mammals is regulated by the noncoding Xist RNA and is associated with the differential acquisition of active and repressive histone modifications, resulting in repression of most genes on one of the two X chromosome homologs. Marsupial mammals exhibit dosage compensation; however, they lack Xist, and the mechanisms conferring epigenetic control of X chromosome dosage compensation remain elusive. Oviparous mammals, the monotremes, have multiple X chromosomes, and it is not clear whether they undergo dosage compensation and whether there is epigenetic dimorphism between homologous pairs in female monotremes. Here, using antibodies against DNA methylation, eight different histone modifications, and HP1, we conduct immunofluorescence on somatic cells of the female Australian marsupial possum Trichosurus vulpecula, the female platypus Ornithorhynchus anatinus, and control mouse cells. The two marsupial X's were different for all epigenetic features tested. In particular, unlike in the mouse, both repressive modifications, H3K9me3 and H4K20Me3, are enriched on one of the X chromosomes, and this is associated with the presence of HP1 and hypomethylation of DNA. Using sequential labeling, we determine that this DNA hypomethylated X correlates with histone marks of inactivity. These results suggest that female marsupials use a repressive histone-mediated inactivation mechanism and that this may represent an ancestral dosage compensation process that differs from eutherians that require Xist transcription and DNA methylation. In comparison to the marsupial, the monotreme exhibited no epigenetic differences between homologous X chromosomes, suggesting the absence of a dosage compensation process comparable to that in therians.

Journal ArticleDOI
TL;DR: It is shown that recombinant MSL2, through its CXC domain, directly binds DNA with low nanomolar affinity, suggesting the existence of an unknown selectivity cofactor in vivo.
Abstract: Dosage compensation in Drosophila melanogaster involves the selective targeting of the male X chromosome by the dosage compensation complex (DCC) and the coordinate, approximately 2-fold activation of most genes. The principles that allow the DCC to distinguish the X chromosome from the autosomes are not understood. Targeting presumably involves DNA sequence elements whose combination or enrichment mark the X chromosome. DNA sequences that characterize 'chromosomal entry sites' or 'high-affinity sites' may serve such a function. However, to date no DNA binding domain that could interpret sequence information has been identified within the subunits of the DCC. Early genetic studies suggested that MSL1 and MSL2 serve to recognize high-affinity sites (HAS) in vivo, but a direct interaction of these DCC subunits with DNA has not been studied. We now show that recombinant MSL2, through its CXC domain, directly binds DNA with low nanomolar affinity. The DNA binding of MSL2 or of an MSL2-MSL1 complex does not discriminate between different sequences in vitro, but in a reporter gene assay in vivo, suggesting the existence of an unknown selectivity cofactor. Reporter gene assays and localization of GFP-fusion proteins confirm the important contribution of the CXC domain for DCC targeting in vivo.

Journal ArticleDOI
TL;DR: The dosage-compensation systems that evolved along with monosomic sex chromosomes exemplify the intricacies of compensating for differences in gene copy number by transcriptional regulation.
Abstract: Diploid genomes are exquisitely balanced systems of gene expression. The dosage-compensation systems that evolved along with monosomic sex chromosomes exemplify the intricacies of compensating for differences in gene copy number by transcriptional regulation.

Journal ArticleDOI
TL;DR: Gene ontology analyses show that among these male-biased genes found on the Z chromosome, more than 20 genes play a role in sex differentiation, corroborate previous studies demonstrating the rather inefficient dosage compensation for Z chromosome in birds and suggest a potential role of non-compensated Z-linked genes in somatic sex differentiation in birds.
Abstract: Considerable progress has been made in our understanding of sex determination and dosage compensation mechanisms in model organisms such as C. elegans, Drosophila and M. musculus. Strikingly, the mechanism involved in sex determination and dosage compensation are very different among these three model organisms. Birds present yet another situation where the heterogametic sex is the female. Sex determination is still poorly understood in birds and few key determinants have so far been identified. In contrast to most other species, dosage compensation of bird sex chromosomal genes appears rather ineffective. By comparing microarrays from microdissected primitive streak from single chicken embryos, we identified a large number of genes differentially expressed between male and female embryos at a very early stage (Hamburger and Hamilton stage 4), long before any sexual differentiation occurs. Most of these genes are located on the Z chromosome, which indicates that dosage compensation is ineffective in early chicken embryos. Gene ontology analyses, using an enhanced annotation tool for Affymetrix probesets of the chicken genome developed in our laboratory (called Manteia), show that among these male-biased genes found on the Z chromosome, more than 20 genes play a role in sex differentiation. These results corroborate previous studies demonstrating the rather inefficient dosage compensation for Z chromosome in birds and show that this sexual dimorphism in gene regulation is observed long before the onset of sexual differentiation. These data also suggest a potential role of non-compensated Z-linked genes in somatic sex differentiation in birds.

Journal ArticleDOI
01 Mar 2010-Placenta
TL;DR: Imprinted paternal X inactivation in rodent extraembryonic tissues would be expected to mitigate the effect of foreign paternal antigens; however, paternal inactivation seems not to occur in the human placenta.

Journal ArticleDOI
TL;DR: It is proposed that the extensive male bias on Z chromosomes is caused by the functional properties of male-adapted genes, which make them unsuitable for high expression in females, and that ZW females are dosage-compensated to a point where they have achieved enough compensation to maintain the integrity of critical networks.

Journal ArticleDOI
TL;DR: The results suggest that overexpression of TBG is responsible for the fat accumulation observed in H2afy-deficient liver, and demonstrate a role for macroH2A1 in regulating homeostasis in a sex-dependent manner, subject to genetic background.
Abstract: Background Vertebrate heterochromatin contains a non-allelic variant of the histone H2A called macroH2A1, which has the characteristic of being three times the size of the canonical H2A. The macroH2A1 C-terminal extension can recruit onto chromatin the poly-ADP-ribose polymerase (PARP)1, which is crucial for DNA repair. This led to the speculation that macroH2A1 could be essential for genome surveillance; however, no experimental evidence supported this hypothesis. Because macroH2A1 has been found to be enriched on the inactive X-chromosome in females, it is thought to play a role in sex chromosome dosage compensation through its ability to regulate gene expression. However, more genetic data are needed to further understand the function of macroH2A1 in mammals.

Journal ArticleDOI
TL;DR: In the tammar wallaby, one X is expressed in all cells, but genes on the other (paternal) X escape inactivation independently and at characteristic frequencies, which appears to be quite distinct from the X chromosome inactivation process in eutherians.
Abstract: X chromosome inactivation is a spectacular example of epigenetic silencing. In order to deduce how this complex system evolved, we examined X inactivation in a model marsupial, the tammar wallaby (Macropus eugenii). In marsupials, X inactivation is known to be paternal, incomplete and tissue-specific, and occurs in the absence of an XIST orthologue. We examined expression of X-borne genes using quantitative PCR, revealing a range of dosage compensation for different loci. To assess the frequency of 1X- or 2X-active fibroblasts, we investigated expression of 32 X-borne genes at the cellular level using RNA-FISH. In female fibroblasts, two-color RNA-FISH showed that genes were coordinately expressed from the same X (active X) in nuclei in which both loci were inactivated. However, loci on the other X escape inactivation independently, with each locus showing a characteristic frequency of 1X-active and 2X-active nuclei, equivalent to stochastic escape. We constructed an activity map of the tammar wallaby inactive X chromosome, which identified no relationship between gene location and extent of inactivation, nor any correlation with the presence or absence of a Y-borne paralog. In the tammar wallaby, one X (presumed to be maternal) is expressed in all cells, but genes on the other (paternal) X escape inactivation independently and at characteristic frequencies. The paternal and incomplete X chromosome inactivation in marsupials, with stochastic escape, appears to be quite distinct from the X chromosome inactivation process in eutherians. We find no evidence for a polar spread of inactivation from an X inactivation center.

Journal ArticleDOI
TL;DR: In this article, a discussion of the normalization procedures for studying gene expression in aneuploid genotypes is presented concerning gene expression and the modulations of gene expression, which can provide insight into the nature of cancerous cells.
Abstract: Aneuploidy involves changes in chromosomal copy number compared with normal euploid genotypes. Studies of gene expression in aneuploids in a variety of species have claimed many different types of responses. Studies of individual genes suggest that there are both structural gene dosage effects and compensation in aneuploids, and that subtle trans-acting effects across the genome are quite prevalent. A discussion is presented concerning the normalization procedures for studying gene expression in aneuploids. A careful documentation of the modulations of gene expression in aneuploids should provide insight into the nature of cancerous cells and the basis of aneuploid syndromes.

Journal ArticleDOI
TL;DR: The data indicate that X chromatin structure is fundamentally different from autosome structure in both sexes, and it is suggested that the differences between the X chromosomes and autosomes in females are a consequence of mechanisms that have evolved to ensure sufficient X chromosome expression in the soma of males.
Abstract: Background X chromosomes are subject to dosage compensation in Drosophila males. Dosage compensation requires cis sequence features of the X chromosome that are present in both sexes by definition and trans acting factors that target chromatin modifying machinery to the X specifically in males. The evolution of this system could result in neutral X chromatin changes that will be apparent in females.